Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Int J Mol Sci ; 24(12)2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37373104

ABSTRACT

Interleukin-7 (IL-7) plays a vital role in the homeostasis of CD4+ and CD8+ T cells. Although IL-7 has been implicated in T helper (Th)1- and Th17-mediated autoinflammatory diseases, its role in Th2-type allergic disorders, such as atopic dermatitis (AD), remains unclear. Thus, to elucidate the effects of IL-7 deficiency on AD development, we generated IL-7-deficient AD-prone mice by backcrossing IL-7 knockout (KO) B6 mice onto the NC/Nga (NC) mouse strain, a model for human AD. As expected, IL-7 KO NC mice displayed defective development of conventional CD4+ and CD8+ T cells compared with wild type (WT) NC mice. However, IL-7 KO NC mice presented with enhanced AD clinical scores, IgE hyperproduction, and increased epidermal thickness compared with WT NC mice. Moreover, IL-7 deficiency decreased Th1, Th17, and IFN-γ-producing CD8+ T cells but increased Th2 cells in the spleen of NC mice, indicating that a reduced Th1/Th2 ratio correlates with severity of AD pathogenesis. Furthermore, significantly more basophils and mast cells infiltrated the skin lesions of IL-7 KO NC mice. Taken together, our findings suggest that IL-7 could be a useful therapeutic target for treating Th2-mediated skin inflammations, such as AD.


Subject(s)
Dermatitis, Atopic , Skin Diseases , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/pathology , Cytokines , Dermatitis, Atopic/drug therapy , Disease Models, Animal , Interleukin-7/genetics , Interleukin-7/metabolism , Skin/pathology , Skin Diseases/pathology , Th2 Cells
2.
Molecules ; 28(15)2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37570749

ABSTRACT

Beta-glucan (ß-glucan) is a natural polysaccharide produced by fungi, bacteria, and plants. Although it has been reported that ß-glucan enhances innate immune memory responses, it is unclear whether different types of ß-glucans display similar immune effects. To address this issue, we employed zymosan (ß-1,3-glycosidic linkage) and pustulan (ß-1,6-glycosidic linkage) to investigate their in vivo effects on innate memory immune responses. We examined the changes of innate memory-related markers in macrophages and natural killer (NK) cells, two immune cell types that display innate memory characteristics, at two different time points (16 h and 7 days) after ß-glucan stimulation. We found that short-term (16 h) zymosan treatment significantly induced macrophages to upregulate IL15 production and increased surface IL15Rα expression on NK cells. In addition, long-term (7 days) zymosan treatment significantly induced macrophages to upregulate the expression of innate memory-related markers (e.g., TNFα, HIF1α, and mTOR) and induced NK cells to express enhanced levels of KLRG1, known as an innate memory-like marker. Our results provide support that zymosan can be an effective adjuvant to promote innate memory immune responses, providing a bridge between innate and adaptive immune cells to enhance various immune responses such as those directed against tumors.


Subject(s)
Interleukin-15 , beta-Glucans , Mice , Animals , Zymosan/pharmacology , Macrophages , beta-Glucans/pharmacology , Killer Cells, Natural , Immunity, Innate
3.
Int J Mol Sci ; 23(23)2022 Dec 05.
Article in English | MEDLINE | ID: mdl-36499642

ABSTRACT

We have previously shown that a deficiency of CD1d-restricted invariant natural killer T (iNKT) cells exacerbates dextran sulfate sodium (DSS)-induced colitis in Yeti mice that exhibit IFNγ-mediated hyper-inflammation. Although iNKT cell-deficiency resulted in reduced Foxp3 expression by mesenteric lymph node (MLN) CD4+ T cells in DSS-treated Yeti mice, the cellular mechanisms that regulate Foxp3 expression by CD25+CD4+ T cells during intestinal inflammation remain unclear. We found that Foxp3-CD25+CD4+ T cells expressing Th1 and Th17 phenotypic hallmarks preferentially expanded in the MLNs of DSS-treated Yeti/CD1d knockout (KO) mice. Moreover, adoptive transfer of Yeti iNKT cells into iNKT cell-deficient Jα18 KO mice effectively suppressed the expansion of MLN Foxp3-CD25+CD4+ T cells during DSS-induced colitis. Interestingly, MLN dendritic cells (DCs) purified from DSS-treated Yeti/CD1d KO mice promoted the differentiation of naive CD4+ T cells into Foxp3-CD25+CD4+ T cells rather than regulatory T (Treg) cells, indicating that MLN DCs might mediate Foxp3+CD25+CD4+ T cell expansion in iNKT cell-sufficient Yeti mice. Furthermore, we showed that Foxp3-CD25+CD4+ T cells were pathogenic in DSS-treated Yeti/CD1d KO mice. Our result suggests that pro-inflammatory DCs and CD1d-restricted iNKT cells play opposing roles in Foxp3 expression by MLN CD25+CD4+ T cells during IFNγ-mediated intestinal inflammation, with potential therapeutic implications.


Subject(s)
Colitis , Dendritic Cells , Natural Killer T-Cells , Animals , Mice , Colitis/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Interferon-gamma/metabolism , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , CD4-Positive T-Lymphocytes/immunology , Interleukin-2 Receptor alpha Subunit , Dendritic Cells/immunology , Dendritic Cells/metabolism
4.
Molecules ; 27(24)2022 Dec 09.
Article in English | MEDLINE | ID: mdl-36557876

ABSTRACT

Although 3-aminopropyl functionalized magnesium phyllosilicate nanoparticles (hereafter aminoclay nanoparticles, ACNs) are well-known nanomaterials employed as drug carriers, their effects on immune cells remain unclear. To address this issue, we explored murine dendritic cells (DCs) as these cells belong to the innate arm of the immune system and function as antigen-presenting cells to elicit adaptive immune responses. We examined the in vitro effects of ACNs on DCs isolated from B6 mice. ACN treatment significantly down-regulated the expression of inflammasome-related markers, including NLRP3, caspase-1, and IL1ß. The ACNs-induced anti-inflammatory DC phenotype was further confirmed by down-regulation of the AKT/mTOR/HIF1α signaling pathway. Such anti-inflammatory effects of ACNs on DCs occurred independently of DC subtypes. To document the effects of ACNs on DCs more clearly, we examined their anti-inflammatory effects on lipopolysaccharide (LPS)-activated DCs. As expected, excessive inflammatory responses (increased mitochondrial ROS and Th1-type cytokines such as IL12 and IL1ß) of LPS-activated DCs were dramatically attenuated by ACN treatment. Furthermore, ACNs down-regulated IFNγ production by antigen-specific CD4+ T cells, which is consistent with a reduced inflammatory phenotype of DCs. Overall, our results provide support for employing ACNs as drug delivery materials with therapeutic potential to control inflammatory disorders.


Subject(s)
Lipopolysaccharides , Nanoparticles , Animals , Mice , Lipopolysaccharides/pharmacology , Cytokines/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/metabolism , Immunity , Dendritic Cells
5.
Int J Mol Sci ; 22(3)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33513946

ABSTRACT

We have previously shown that CD1d-restricted iNKT cells suppress dysregulated IFNγ expression and intestinal inflammation in Yeti mice on the C57BL/6 background. Since type 3 innate lymphoid cells (ILC3s) in mesenteric lymph nodes (MLN) protect against intestinal inflammation in a CD1d-associated manner, we investigated whether crosstalk between iNKT cells and MLN ILC3s controls IFNγ-mediated intestinal inflammation in Yeti mice. We found that Yeti mice display increased levels of ILC3s and that iNKT cell deficiency in Yeti/CD1d KO mice decreases levels of IL22-producing ILC3s during DSS-induced colitis. This finding indicates that iNKT cells and ILC3s cooperate to regulate intestinal inflammation in Yeti mice. Yeti iNKT cells displayed a pronounced anti-inflammatory (IL4- or IL9-producing) phenotype during colitis. Their adoptive transfer to iNKT cell-deficient animals induced a significant increase in IL22 production by ILC3s, indicating that crosstalk between iNKT cells and ILC3s plays a critical role in modulating colitis in Yeti mice. Moreover, we showed that the IL9-producing subset of iNKT cells potently enhances IL22-producing ILC3s in vivo. Taken together, our results identify a central role of the iNKT cell-ILC3 axis in ameliorating IFNγ-mediated intestinal inflammation.


Subject(s)
Antigens, CD1d/genetics , Inflammation/genetics , Interferon-gamma/genetics , Interleukins/genetics , Lymphocytes/metabolism , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Dextran Sulfate/toxicity , Humans , Immunity, Innate/genetics , Inflammation/immunology , Inflammation/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestines/pathology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphocytes/drug effects , Lymphocytes/immunology , Mice , Mice, Knockout , Natural Killer T-Cells/immunology , Natural Killer T-Cells/pathology , Splanchnic Circulation/genetics , Interleukin-22
6.
Int J Mol Sci ; 22(6)2021 Mar 16.
Article in English | MEDLINE | ID: mdl-33809795

ABSTRACT

We previously showed that ubiquitous overexpression of the chromatin remodeling factor SWItch3-related gene (SRG3) promotes M2 macrophage differentiation, resulting in anti-inflammatory responses in the experimental autoimmune encephalomyelitis model of multiple sclerosis. Since hepatic macrophages are responsible for sepsis-induced liver injury, we investigated herein the capacity of transgenic SRG3 overexpression (SRG3ß-actin mice) to modulate sepsis in mice exposed to lipopolysaccharide (LPS) plus d-galactosamine (d-GalN). Our results demonstrated that ubiquitous SRG3 overexpression significantly protects mice from LPS/d-GalN-induced lethality mediated by hepatic M1 macrophages. These protective effects of SRG3 overexpression correlated with the phenotypic conversion of hepatic macrophages from an M1 toward an M2 phenotype. Furthermore, SRG3ß-actin mice had decreased numbers and activation of natural killer (NK) cells but not natural killer T (NKT) cells in the liver during sepsis, indicating that SRG3 overexpression might contribute to cross-talk between NK cells and macrophages in the liver. Finally, we demonstrated that NKT cell-deficient CD1d KO/SRG3ß-actin mice are protected from LPS/d-GalN-induced sepsis, indicating that NKT cells are dispensable for SRG3-mediated sepsis suppression. Taken together, our findings provide strong evidence that SRG3 overexpression may serve as a therapeutic approach to control overwhelming inflammatory diseases such as sepsis.


Subject(s)
Chromatin/metabolism , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Liver/pathology , Macrophages/metabolism , Natural Killer T-Cells/metabolism , Sepsis/chemically induced , Sepsis/prevention & control , Transcription Factors/metabolism , Actins/genetics , Animals , Chromatin Assembly and Disassembly , Dendritic Cells/metabolism , Galactosamine , Inflammation Mediators/metabolism , Lipopolysaccharides , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , Protective Agents/metabolism , Sepsis/immunology , Sepsis/pathology , Severity of Illness Index
7.
Int J Mol Sci ; 22(4)2021 Feb 04.
Article in English | MEDLINE | ID: mdl-33557054

ABSTRACT

The SWItch (SWI)3-related gene (SRG3) product, a SWI/Sucrose Non-Fermenting (SNF) chromatin remodeling subunit, plays a critical role in regulating immune responses. We have previously shown that ubiquitous SRG3 overexpression attenuates the progression of Th1/Th17-mediated experimental autoimmune encephalomyelitis. However, it is unclear whether SRG3 overexpression can affect the pathogenesis of inflammatory skin diseases such as atopic dermatitis (AD), a Th2-type immune disorder. Thus, to elucidate the effects of SRG3 overexpression in AD development, we bred NC/Nga (NC) mice with transgenic mice where SRG3 expression is driven by the ß-actin promoter (SRG3ß-actin mice). We found that SRG3ß-actin NC mice exhibit increased AD development (e.g., a higher clinical score, immunoglobulin E (IgE) hyperproduction, and an increased number of infiltrated mast cells and basophils in skin lesions) compared with wild-type NC mice. Moreover, the severity of AD pathogenesis in SRG3ß-actin NC mice correlated with expansion of interleukin 4 (IL4)-producing basophils and mast cells, and M2 macrophages. Furthermore, this accelerated AD development is strongly associated with Treg cell suppression. Collectively, our results have identified that modulation of SRG3 function can be applied as one of the options to control AD pathogenesis.


Subject(s)
Chromatin Assembly and Disassembly , Dermatitis, Atopic/etiology , Gene Expression , Th2 Cells/immunology , Th2 Cells/metabolism , Transcription Factors/genetics , Actins/metabolism , Animals , Biopsy , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dermatitis, Atopic/diagnosis , Dermatitis, Atopic/metabolism , Disease Models, Animal , Disease Susceptibility , Immunity, Cellular , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Transgenic , Severity of Illness Index
8.
J Nanobiotechnology ; 18(1): 133, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32933548

ABSTRACT

BACKGROUND: Infliximab (IFX), a TNF-α blocking chimeric monoclonal antibody, induces clinical response and mucosal healing in patients with inflammatory bowel disease (IBD). However, systemic administration of this agent causes unwanted side effects. Oral delivery of antibody therapeutics might be an effective treatment strategy for IBD compared to intravenous administration. RESULTS: All three carriers had a high encapsulation efficiency, narrow size distribution, and minimal systemic exposure. There was a higher interaction between nanocomposite carriers and monocytes compared to lymphocytes in the PBMC of IBD patients. Orally administered nanocomposite carriers targeted to inflamed colitis minimized systemic exposure. All IFX delivery formulations with nanocomposite carriers had a significantly less colitis-induced body weight loss, colon shortening and histomorphological score, compared to the DSS-treated group. AC-IFX-L and EAC-IFX-L groups showed significantly higher improvement of the disease activity index, compared to the DSS-treated group. In addition, AC-IFX-L and EAC-IFX-L alleviated pro-inflammatory cytokine expressions (Tnfa, Il1b, and Il17). CONCLUSION: We present orally administered antibody delivery systems which improved efficacy in murine colitis while reducing systemic exposure. These oral delivery systems suggest a promising therapeutic approach for treating IBD.


Subject(s)
Colitis/drug therapy , Drug Delivery Systems/methods , Infliximab/pharmacology , Nanocomposites/administration & dosage , Nanocomposites/chemistry , Administration, Oral , Animals , Antibodies, Monoclonal , Colitis/pathology , Colon/pathology , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology , Leukocytes, Mononuclear , Liposomes , Lymphocytes , Mice , Mice, Inbred C57BL , Particle Size , Tumor Necrosis Factor-alpha/drug effects
9.
Mediators Inflamm ; 2019: 3540974, 2019.
Article in English | MEDLINE | ID: mdl-30766446

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder which is caused by a mutation of the huntingtin (HTT) gene. Although the pathogenesis of HD has been associated with inflammatory responses, if and how the immune system contributes to the onset of HD is largely unknown. Invariant natural killer T (iNKT) cells are a group of innate-like regulatory T lymphocytes that can rapidly produce various cytokines such as IFNγ and IL4 upon stimulation with the glycolipid α-galactosylceramide (α-GalCer). By employing both R6/2 Tg mice (murine HD model) and Jα18 KO mice (deficient in iNKT cells), we investigated whether alterations of iNKT cells affect the development of HD in R6/2 Tg mice. We found that Jα18 KO R6/2 Tg mice showed disease progression comparable to R6/2 Tg mice, indicating that the absence of iNKT cells did not have any significant effects on HD development. However, repeated activation of iNKT cells with α-GalCer facilitated HD progression in R6/2 Tg mice, and this was associated with increased infiltration of iNKT cells in the brain. Taken together, our results demonstrate that repeated α-GalCer treatment of R6/2 Tg mice accelerates HD progression, suggesting that immune activation can affect the severity of HD pathogenesis.


Subject(s)
Huntington Disease/immunology , Lymphocyte Activation , Natural Killer T-Cells/immunology , Animals , Brain/metabolism , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Galactosylceramides/chemistry , Genotype , Leukocytes/metabolism , Mice , Mice, Knockout
10.
Environ Geochem Health ; 41(4): 1723-1728, 2019 Aug.
Article in English | MEDLINE | ID: mdl-28455818

ABSTRACT

Lipid derivatization technology-mediated fatty acid profiling studies have been suggested to dissect the contents of lipids in white fat and brown fat tissue. The focus of this study is to profile fatty acid lipidomics in brown adipose tissue and white adipose tissue of mice by derivatizing their lipids into fatty acid methyl esters via in situ transmethylation using a rice husk-derived biochar as porous media. The in situ transmethylation using biochar is advantageous in biological analysis because there was no loss of samples inevitably occurring in the loss of lipid in solvent extraction and purification steps.


Subject(s)
Adipose Tissue, Brown/chemistry , Adipose Tissue, White/chemistry , Charcoal/chemistry , Fatty Acids/analysis , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Fatty Acids/chemistry , Female , Lipids/chemistry , Male , Methylation , Mice, Inbred C57BL
11.
EMBO J ; 33(3): 217-28, 2014 Feb 03.
Article in English | MEDLINE | ID: mdl-24442639

ABSTRACT

TopBP1 was initially identified as a topoisomerase II-ß-binding protein and it plays roles in DNA replication and repair. We found that TopBP1 is expressed at high levels in lymphoid tissues and is essential for early lymphocyte development. Specific abrogation of TopBP1 expression resulted in transitional blocks during early lymphocyte development. These defects were, in major part, due to aberrant V(D)J rearrangements in pro-B cells, double-negative and double-positive thymocytes. We also show that TopBP1 was located at sites of V(D)J rearrangement. In TopBP1-deficient cells, γ-H2AX foci were found to be increased. In addition, greater amount of γ-H2AX product was precipitated from the regions where TopBP1 was localized than from controls, indicating that TopBP1 deficiency results in inefficient DNA double-strand break repair. The developmental defects were rescued by introducing functional TCR αß transgenes. Our data demonstrate a novel role for TopBP1 as a crucial factor in V(D)J rearrangement during the development of B, T and iNKT cells.


Subject(s)
Carrier Proteins/genetics , DNA Repair , DNA/genetics , Lymphocytes/physiology , V(D)J Recombination/immunology , Animals , Carrier Proteins/immunology , Carrier Proteins/metabolism , Chromatin Immunoprecipitation , DNA Breaks, Double-Stranded , DNA Damage , Gene Expression , Histones/genetics , Histones/metabolism , Lymphocytes/immunology , Mice , Mice, Knockout , Natural Killer T-Cells/immunology , Natural Killer T-Cells/physiology , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/physiology , Sequence Deletion , Specific Pathogen-Free Organisms , Transgenes , V(D)J Recombination/genetics
12.
BMC Complement Altern Med ; 18(1): 218, 2018 Jul 18.
Article in English | MEDLINE | ID: mdl-30021579

ABSTRACT

BACKGROUND: The polysaccharide component of Angelica gigas induces immuno-stimulatory effects on innate immune cells. However, it is unclear whether A. gigas' adjuvant activity on the immune system can elicit anti-cancer responses. METHODS: A water-soluble immuno-stimulatory component of A. gigas was prepared. How this ISAg modulated the activation of innate immune cells such as dendritic cells (DCs) was examined. ISAg-induced cytotoxic activity via natural killer (NK) and NKT cells was also tested using a tumor-bearing mouse model. RESULTS: ISAg treatment induced nitric oxide (NO) production and cytokine gene expression involved in innate immune responses. ISAg activated macrophages and DCs to secrete cytokine IL-12, through the TLR4 signaling pathway. IL-12 plays a crucial role in ISAg-mediated NK and NKT cell activation. Thus, the anti-cancer activity of NK and NKT cells induced ISAg-mediated cytotoxicity of B16 melanoma cells in mice. CONCLUSIONS: These results indicated that the natural ingredient, ISAg, has adjuvant activity to induce strong anti-cancer activity of NK and NKT cells in vivo.


Subject(s)
Angelica/chemistry , Antineoplastic Agents/pharmacology , Immunity, Innate/drug effects , Killer Cells, Natural , Natural Killer T-Cells , Plant Extracts/pharmacology , Animals , Cell Line, Tumor , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Mice , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , Polysaccharides/pharmacology , RAW 264.7 Cells , Xenograft Model Antitumor Assays
13.
BMC Complement Altern Med ; 17(1): 448, 2017 Sep 06.
Article in English | MEDLINE | ID: mdl-28877696

ABSTRACT

BACKGROUND: Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders that are mediated by pathogenic Th1 and Th17 cells. Previous studies have demonstrated that taheebo water extract (TWE) derived from Tabebuia avellanedae Lorentz ex Griseb., as folk remedy, has been used to treat various inflammatory diseases. Although TWE has been previously shown to display anti-inflammatory activities, the in vivo effects of TWE on mucosal immune responses remain unclear. METHODS: We examined the anti-inflammatory effects of TWE on innate immune cells such as dendritic cells (DCs) and macrophages and also on the differentiation of T helper cells. Lastly, adopting a method for dextran sulfate sodium (DSS)-induced colitis, we investigated whether the oral administration of TWE can modulate mucosal inflammatory responses. RESULTS: We found that TWE could activate DCs to produce immunosuppressive IL10 and polarize macrophages toward an anti-inflammatory phenotype in the mesenteric lymph node (MLN). Such alterations in DCs and macrophages resulted in a significant increase in anti-inflammatory Th2 and Foxp3+ Treg cells and a dramatic decrease in pro-inflammatory Th1 and Th17 cells in the MLN. Upon induction of colitis with DSS treatment, TWE significantly reduced the clinical symptoms, including body weight loss and colonic tissue inflammation, by up-regulating type II T helper immune responses. CONCLUSIONS: Taken together, these data suggest that TWE is an excellent natural product with therapeutic effects to help improve inflammatory disorders such as colitis.


Subject(s)
Colitis , Plant Extracts , Th2 Cells/drug effects , Th2 Cells/immunology , Administration, Oral , Animals , Colitis/chemically induced , Colitis/drug therapy , Colitis/immunology , Cytokines/immunology , Cytokines/metabolism , Dextran Sulfate , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
14.
Clin Immunol ; 157(2): 156-65, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25725428

ABSTRACT

Both the thymus (T) and bone (B) are necessary hematopoietic niches in adult humans. We previously showed that co-transplantation of human fetal T and B tissues into neonatal immunodeficient NOD/SCID IL2Rγ(null) (NSG, N) mice facilitated hematopoiesis. However, transplantation into neonatal mice resulted in high frequency of early death, making it unrealistic for repetitive experiments. In this study, young adult N mice were pre-engrafted with T and B, T alone, B alone or no tissues. The animals were irradiated and injected with autologous fetal liver (FL)-derived CD34(+) cells (34). The resultant mice were TB34N, T34N, B34N and 34N, respectively, and challenged with T cell dependent antigens (Ags). The humanized TB34N mice showed best performance of these mouse models in many aspects resembling the adult human Ag-experienced spleen. The TB34N mice exhibited better hematopoietic reconstitution; balanced development of T- and B-cell, and common progenitor cells; follicular lymphoid structures with a functional germinal center (GC) enriched with follicular dendritic cells (FDCs) and plasma cells (PCs); secretion of hIgG in the sera in response to Ags at comparable levels to those of human; derivations of hIgG mAb-secreting hybridoma clones. Collectively, the humanized TB34N mice could develop an adaptive immunity that was capable of producing Ag-specific hIgG at a significant level via class switching. This unprecedented TB34N platform in humanized mice would be useful in dissecting human immunity, for generating human Abs and clinical applications.


Subject(s)
Adaptive Immunity/immunology , Antibodies/immunology , Antigens, CD34/metabolism , Bone Transplantation , Fetal Tissue Transplantation , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Spleen/immunology , Thymus Gland/transplantation , Animals , Antibody Formation , Hematopoiesis , Heterografts , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Interleukin Receptor Common gamma Subunit/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Spleen/pathology
15.
Biochem Biophys Res Commun ; 461(1): 86-94, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25858316

ABSTRACT

The inflammatory cytokine IL32γ acts on dendritic cells (DCs) to produce IL12 and IL6, which are involved in the differentiation of Th1 and Th17 cells. Natural killer (NK) and NKT cells play important roles in IL12-mediated adaptive immune responses, such as antitumor immunity. Herein we demonstrate the effect of IL32γ on the activation of NK and NKT cells. Upon IL32γ stimulation, splenic NK and NKT cells could be activated, and this activation was dependent on both IL12 and DCs, which was confirmed by using IL12p35 knockout and CD11c-diphtheria toxin receptor transgenic mouse models. Furthermore, IL32γ could induce the production of proinflammatory cytokines by NKDCs, a subset of DCs expressing NK cell markers, known to enhance NKT cell function. Unlike conventional DCs, NKDCs produced IFNγ and TNFα rather than IL12 upon stimulation with IL32γ. Taken together, IL32γ will be useful as an adjuvant to boost the cytotoxicities of NK and NKT cells that play critical roles in antitumor immunity.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate/immunology , Interferon-gamma/immunology , Interleukin-12/immunology , Interleukins/immunology , Killer Cells, Natural/immunology , Natural Killer T-Cells/immunology , Animals , Cell Communication/immunology , Cells, Cultured , Mice , Mice, Inbred C57BL
16.
Biochem Biophys Res Commun ; 443(2): 413-21, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24309101

ABSTRACT

Bacillus subtilis-derived poly-γ-glutamic acid (γPGA) stimulates dendritic cells (DCs) to produce IL12, leading to CD4(+) T cell differentiation toward the Th1 phenotype, but DCs consist of heterogeneous subpopulations with a variety of immune functions. Among these, natural killer dendritic cells (NKDCs) play an important role in anti-tumor immune responses. Herein, we demonstrate the role of NKDCs in γPGA-meditated anti-tumor immune responses. NK1.1(+) CD11c(+) NKDCs were stimulated upon γPGA stimulation in vitro and in vivo to up-regulate lymphocyte activation markers, MHC class I and II, and co-stimulatory molecules. In particular, NKDCs were activated by γPGA to produce IFNγ and TNFα, like NK cells, as well as IL12, like DCs, implying that NKDCs have unique and multifunctional roles. Importantly, NKDCs stimulated by γPGA conferred stronger anti-tumor effects in mice and showed increased cytotoxicity against various tumor cell lines in vitro. In conclusion, NKDCs are one of the key players in anti-tumor immunity induced by γPGA.


Subject(s)
Bacillus subtilis/metabolism , Dendritic Cells/immunology , Immunity, Cellular/immunology , Immunologic Factors/pharmacology , Killer Cells, Natural/immunology , Polyglutamic Acid/analogs & derivatives , Animals , Cells, Cultured , Cytokines/immunology , Dendritic Cells/drug effects , Immunity, Cellular/drug effects , Immunologic Factors/isolation & purification , Immunologic Factors/metabolism , Killer Cells, Natural/drug effects , Mice , Mice, Inbred C57BL , Polyglutamic Acid/isolation & purification , Polyglutamic Acid/metabolism , Polyglutamic Acid/pharmacology
17.
Exp Dermatol ; 22(8): 561-3, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23879818

ABSTRACT

Bacillus subtilis-derived poly-γ-glutamic acid (γPGA) has demonstrated adjuvant activity in promoting Th1/Th17 cell differentiation. Here, the NC/Nga (NC) mouse model was used to determine whether γPGA modulates the outcome of atopic dermatitis (AD), which is known to be a Th2-biased immune disease. We found that oral administration of γPGA dramatically reduced the development of AD in NC mice. Antigen-presenting cells activated with γPGA produced pro-inflammatory cytokines, such as IL12/23 and IFNγ, which, in turn, induced the differentiation of Th1 and Th17 cells. Concomitantly, Th2 responses, such as high levels of serum IgE, were dramatically decreased. Furthermore, in vivo γPGA treatment altered several cellular components of allergic reactions, such as mast cells and eosinophils. Taken together, our results strongly demonstrate that in vivo treatment with γPGA at early time points can prevent the development of AD in NC mice and suggest that γPGA may have therapeutic applications for human AD.


Subject(s)
Dermatitis, Atopic/prevention & control , Gene Expression Regulation , Polyglutamic Acid/analogs & derivatives , Administration, Oral , Animals , Antigen-Presenting Cells/cytology , CD3 Complex/metabolism , Cell Differentiation , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Eosinophils/cytology , Immunoglobulin E/chemistry , Inflammation , Male , Mast Cells/cytology , Mice , Polyglutamic Acid/administration & dosage , Th1 Cells/cytology , Th17 Cells/cytology
18.
Front Immunol ; 14: 1343718, 2023.
Article in English | MEDLINE | ID: mdl-38274786

ABSTRACT

Invariant natural killer T (iNKT) cells, a subset of unconventional T cells that recognize glycolipid antigens in a CD1d-dependent manner, are crucial in regulating diverse immune responses such as autoimmunity. By engaging with CD1d-expressing non-immune cells (such as intestinal epithelial cells and enterochromaffin cells) and immune cells (such as type 3 innate lymphoid cells, B cells, monocytes and macrophages), iNKT cells contribute to the maintenance of immune homeostasis in the intestine. In this review, we discuss the impact of iNKT cells and CD1d in the regulation of intestinal inflammation, examining both cellular and molecular factors with the potential to influence the functions of iNKT cells in inflammatory bowel diseases such as Crohn's disease and ulcerative colitis.


Subject(s)
Natural Killer T-Cells , Humans , Immunity, Innate , Lymphocytes , Macrophages , Inflammation
19.
J Immunol ; 184(10): 5589-94, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20375305

ABSTRACT

NKT cells are considered to be innate-like regulatory cells. However, their regulatory functions in adaptive immune responses have not been studied in detail. In this study, we investigated the immunoregulatory functions of NKT cells during the secondary phase of an Ag-specific CD4(+) T cell response. When compared with OVA-specific effector CD4(+) T cells adoptively transferred into NKT cell-deficient naive CD1d(-/-) mice, the same T cells transferred into naive CD1d(+/-) mice exhibited substantially stronger immune responses on OVA challenge. The enhanced immune response of the transferred CD4(+) T cells in the presence of NKT cells correlated with an increase in their proliferation in vivo. In addition, T cells transferred into CD1d(+/-) recipients showed enhanced cytokine productions relative to T cells in CD1d(-/-) recipients. To elucidate the physiological relevance of the regulatory role of NKT cells in a disease setting, OVA-specific asthma was induced in recipient mice after adoptive transfer of OVA-specific CD4(+) T cells. CD1d(+/-) recipients showed stronger asthmatic phenotypes in all indications when compared with CD1d(-/-) recipients. Taken together, these results suggest that NKT cells are critical for the regulation of Ag-specific, conventional CD4(+) T cells during the secondary phase of an adaptive immune response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/immunology , Immunization, Secondary , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Adoptive Transfer , Amino Acid Sequence , Animals , Asthma/immunology , Asthma/metabolism , Asthma/pathology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/transplantation , Cell Proliferation , Cells, Cultured , Cytokines/biosynthesis , Disease Models, Animal , Immunity, Cellular/genetics , Immunization, Secondary/methods , Lymphocyte Activation/immunology , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Natural Killer T-Cells/pathology , Ovalbumin/administration & dosage , Ovalbumin/immunology
20.
J Immunol ; 185(3): 1476-84, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20581146

ABSTRACT

The functional role of IL-12 in rheumatoid arthritis is controversial. Moreover, whether IL-12 contributes to regulation of Ab-induced joint inflammation remains unclear. To address these issues, we explored the functional roles of IL-12 in Ab-induced arthritis using the K/BxN serum transfer model. IL-12p35(-/-) and IL-12Rbeta(2)(-/-) mice were resistant to the development of arthritis. Injection of K/BxN serum into IL-12p40-yellow fluorescence protein reporter (yet40) mice induced CD11b(+) cells, CD11c(+) cells, and Gr-1(+) granulocytes to produce IL-12p40 in the joints. The levels of IFN-gamma, IL-4, and IL-6 production were lower in joint tissues of IL-12p35(-/-) and IL-12Rbeta(2)(-/-) mice than in B6 mice, whereas levels of TGF-beta expression were higher. Administering IL-12p35(-/-) mice rIL-12 or IFN-gamma restored joint inflammation and suppressed TGF-beta production in joint tissues. Moreover, administering neutralizing anti-TGF-beta mAb enhanced joint inflammation. Among the immune cells that infiltrated joint tissues during Ab-induced arthritis, NKT cells expressed IL-12beta(2) receptors. Furthermore, the adoptive transfer of splenocytes from B6 or Gr-1(+) granulocyte-depleted mice restored joint inflammation in IL-12Rbeta(2)(-/-) mice as much as in B6 mice, whereas splenocytes from Jalpha18(-/-) mice did not. These findings indicate that signals via IL-12beta(2) receptors on NKT cells play a critical role in the development of Ab-induced arthritis. The IL-12p35/IFN-gamma axis promotes Ab-induced joint inflammation by activating NKT cells and suppressing TGF-beta, which may provide novel information for the development of new therapeutic strategies for the inhibition of rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Immune Sera/administration & dosage , Interleukin-12 Subunit p35/physiology , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Transforming Growth Factor beta/antagonists & inhibitors , Amino Acid Sequence , Animals , Antibodies, Monoclonal/administration & dosage , Arthritis, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Knock-In Techniques , Immune Complex Diseases/immunology , Immune Complex Diseases/metabolism , Immune Complex Diseases/pathology , Immune Sera/blood , Inflammation Mediators/administration & dosage , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/physiology , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology , Receptors, Interleukin-12/deficiency , Receptors, Interleukin-12/genetics , Receptors, Interleukin-12/physiology , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/immunology
SELECTION OF CITATIONS
SEARCH DETAIL